Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Environ Toxicol ; 39(3): 1119-1128, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37853848

ABSTRACT

We recently reported that arsenic caused insulin resistance in differentiated human neuroblastoma SH-SY5Y cells. Herein, we further investigated the effects of sodium arsenite on IGF-1 signaling, which shares downstream signaling with insulin. A time-course experiment revealed that sodium arsenite began to decrease IGF-1-stimulated Akt phosphorylation on Day 3 after treatment, indicating that prolonged sodium arsenite treatment disrupted the neuronal IGF-1 response. Additionally, sodium arsenite decreased IGF-1-stimulated tyrosine phosphorylation of the IGF-1 receptor ß (IGF-1Rß) and its downstream target, insulin receptor substrate 1 (IRS1). These results suggested that sodium arsenite impaired the intrinsic tyrosine kinase activity of IGF-1Rß, ultimately resulting in a reduction in tyrosine-phosphorylated IRS1. Sodium arsenite also reduced IGF-1 stimulated tyrosine phosphorylation of insulin receptor ß (IRß), indicating the potential inhibition of IGF-1R/IR crosstalk by sodium arsenite. Interestingly, sodium arsenite also induced neurite shortening at the same concentrations that caused IGF-1 signaling impairment. A 24-h IGF-1 treatment partially rescued neurite shortening caused by sodium arsenite. Moreover, the reduction in Akt phosphorylation by sodium arsenite was attenuated by IGF-1. Inhibition of PI3K/Akt by LY294002 diminished the protective effects of IGF-1 against sodium arsenite-induced neurite retraction. Together, our findings suggested that sodium arsenite-impaired IGF-1 signaling, leading to neurite shortening through IGF-1/PI3K/Akt.


Subject(s)
Arsenic , Arsenites , Neuroblastoma , Sodium Compounds , Humans , Proto-Oncogene Proteins c-akt/metabolism , Insulin-Like Growth Factor I , Phosphatidylinositol 3-Kinases/metabolism , Neurites/metabolism , Phosphorylation , Tyrosine/metabolism , Tyrosine/pharmacology
2.
Heliyon ; 9(3): e14385, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36925548

ABSTRACT

We recently reported that arsenic disrupted neuronal insulin signaling. Here, we further investigated the effect of arsenic on insulin receptor substrate (IRS) proteins, which are crucial downstream signaling molecules of insulin in differentiated human neuroblastoma SH-SY5Y cells. We also found that prolonged arsenic treatment accelerated the migration of IRS1 and IRS2 on SDS-PAGE. Treatment with phosphatases abolished the arsenic-induced increased mobility of IRS, suggesting that the electrophoretic mobility shift of IRS on SDS-PAGE by arsenic was phosphorylation-dependent. By using label-free mass spectrometry, the phosphorylation sites of IRS1 were found to be S24, S345, S636, T774, S1057, S1058, and S1070, while those of IRS2 were at S645, Y653, T657, S665, S667, S669, S672, S915, and S1203, which were at least 2-fold lower than found in the control. These findings indicated a global hypophosphorylation of IRS proteins after prolonged arsenic treatment. In addition, four novel phosphorylation sites were identified on IRS1 (T774, S1057, S1058, and S1070), with another two on IRS2 (S665 and S667). As basal IRS phosphorylation plays an important role in insulin signaling, the reduction of IRS phosphorylation on multiple residues may underlie arsenic-impaired insulin signaling in neurons.

3.
Asian Pac J Cancer Prev ; 24(2): 425-434, 2023 Feb 01.
Article in English | MEDLINE | ID: mdl-36853289

ABSTRACT

OBJECTIVE: Cholangiocarcinoma (CCA) is a cancer of the bile duct with a poor prognosis. The present study examined the ability of curcumin to sensitize apoptosis in the TNF-related apoptosis-inducing ligand (TRAIL)-resistant CCA cell lines of HuCCA-1 and KKU-213A. METHODS: Apoptosis was measured using a TUNEL assay. Protein expression was determined by immunoblotting. Membrane death receptor 5 (DR5) was detected by flow cytometry. Protein complex was examined by co-immunoprecipitation. RESULT: Curcumin potentiated TRAIL-induced apoptosis in both cell lines, indicating the sensitization to TRAIL-induced apoptosis by curcumin. Additionally, curcumin increased DR5 expression and membrane localization; however, the curcumin/TRAIL combination did not result in further increases in DR5 expression and membrane localization in either cell line. Moreover, the curcumin/TRAIL combination reduced DR5/decoy receptor 2 (DcR2) complexes in both cell lines, suggesting that curcumin may enhance TRAIL-induced apoptosis by disrupting DR5/DcR2 interaction. In addition, levels of the anti-apoptotic complex DR5/ DDX3/GSK3ß were reduced by the curcumin/TRAIL combination in HuCCA-1 but not in KKU-213A cells. This study also demonstrated that the DR5/DcR2 and DR5/DDX3/GSK3ß complexes could be observed under basal conditions, suggesting that these anti-apoptotic complexes may contribute to TRAIL-resistant phenotypes in both cell lines. Pretreatment with the antioxidant N-acetylcysteine attenuated curcumin-enhanced apoptosis by TRAIL, indicating that curcumin sensitized TRAIL-induced apoptosis through an oxidative stress-dependent mechanism. CONCLUSION: The present study demonstrates the potential of using curcumin in combination with TRAIL to yield better TRAIL therapy outcomes in TRAIL-resistant CCA.


Subject(s)
Bile Duct Neoplasms , Cholangiocarcinoma , Curcumin , Humans , Apoptosis , Bile Duct Neoplasms/drug therapy , Bile Ducts, Intrahepatic , Cholangiocarcinoma/drug therapy , Curcumin/pharmacology , Glycogen Synthase Kinase 3 beta , TNF-Related Apoptosis-Inducing Ligand/pharmacology
4.
Environ Toxicol Pharmacol ; 95: 103974, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36089238

ABSTRACT

Herein, we examined whether prolonged arsenic exposure altered tau phosphorylation in the brain of Sprague Dawley rats expressing endogenous wild-type tau. The results showed that daily intraperitoneal injections of 2.5 mg/kg BW sodium arsenite over 28 days caused arsenic accumulation in the rat brain. Interestingly, we found an increase in tau phosphorylation at the Tau 1 region (189-207) and S202 in the hippocampus, S404 in the cerebral cortex, and S396 and S404 in the cerebellum of arsenic-treated rats. Additionally, arsenic increased active ERK1/2 phosphorylation at T202/Y204 in the hippocampus, cerebral cortex, and cerebellum. Meanwhile, we detected increasing active JNK phosphorylation at T183/Y185 in the hippocampus and cerebellum. Moreover, p35, a neuron-specific activator of CDK5, was also elevated in the cerebellum of arsenic-treated rats, suggesting that CDK5 activity may be increased by arsenic. These results suggested that arsenic may induce tau phosphorylation through the activation of tau kinases, ERK1/2, JNK, and CDK5. Together, the findings from this study demonstrated that prolonged arsenic exposure is implicated in neurodegeneration by promoting tau phosphorylation in the rat brain and points toward a possible prevention strategy against neurodegeneration induced by environmental arsenic exposure.


Subject(s)
Arsenic , tau Proteins , Animals , Arsenic/toxicity , Brain/metabolism , Phosphorylation , Rats , Rats, Sprague-Dawley , tau Proteins/metabolism
5.
Vet Med Sci ; 8(1): 69-84, 2022 01.
Article in English | MEDLINE | ID: mdl-34882994

ABSTRACT

BACKGROUND: The anti-cancer effects of Gynura procumbens leaves extract (GPE) have been reported in various human cancers. However, the anti-cancer effects and molecular mechanisms of this extract on canine mammary cancer (CMC) have not yet been elucidated. OBJECTIVES: The main goal of this study was to investigate the anti-cancer properties of GPE against two CMC cell lines (CHMp-13a and CHMp-5b). METHODS: The GP leaves were extracted with 80% ethanol. Anti-cancer potentials of GPE on CHMp-13a and CHMp-5b cancer cell lines using dimethyl-2-thiazolyl-2,5-diphenyl-2H-tetrazolium bromide (MTT), wound healing, transwell migration, and caspase 3/7 activity assays were evaluated. The mRNA expression levels of two oncogenes: epidermal growth factor receptor (EGFR) and twist family bHLH transcription factor 1 (TWIST) and one tumour suppressor gene: phosphatase and tensin homolog (PTEN) in these cell lines were determined by quantitative real-time PCR (qRT-PCR). In addition, The EGFR and PTEN protein levels as well as protein kinase B (AKT) and extracellular signal-regulated kinase 1/2 (ERK1/2) phosphorylation levels expression were also evaluated by western blot analysis. RESULTS: The results showed that GPE caused a significant concentration- and time-dependent reduction in cell proliferation of both CHMp-13a and CHMp-5b cells, detected by MTT assays. This extract also significantly suppressed cancer cell migration in both cell lines, tested by wound healing and transwell migration assays. Additionally, the increase in caspase 3/7 activity observed in both CMC cell treated with GPE suggests that GPE induced caspase 3/7 dependent apoptosis. Moreover, GPE significantly decreased EGFR mRNA and protein expression levels compared to control in both cell lines in a dose-dependent manner. CONCLUSION: These findings emphasized that GPE has an in vitro anti-cancer activity against CMC by inhibiting EGFR signalling pathway. Thus, GPE may serve as an alternative therapy in CMC with high EGFR expression.


Subject(s)
Breast Neoplasms , Dog Diseases , Animals , Breast Neoplasms/veterinary , Cell Line , Cell Proliferation , Dogs , Female , Plant Extracts/pharmacology , Plant Leaves
6.
Toxicol Lett ; 349: 40-50, 2021 Oct 01.
Article in English | MEDLINE | ID: mdl-34118311

ABSTRACT

Previously, we reported that prolonged arsenic exposure impaired neuronal insulin signaling. Here we have further identified novel molecular mechanisms underlying neuronal insulin signaling impairment by arsenic. Arsenic treatment altered insulin dose-response curve and reduced maximum insulin response in differentiated human neuroblastoma SH-SY5Y cells, suggesting that arsenic hindered neuronal insulin signaling in a non-competitive like manner. Mechanistically, arsenic suppressed insulin receptor (IR) kinase activity, as witnessed by a decreased insulin-activated autophosphorylation of IR at Y1150/1151. Arsenic decreased the level of insulin receptor substrate 1 (IRS1) but increased the protein ratio between PI3K regulatory subunit, p85, and PI3K catalytic subunit, p110. Interestingly, co-immunoprecipitation demonstrated that arsenic did not alter a level of PI3K-p110/PI3K-p85 complex while increased PI3K-p85 levels in a PI3K-p110 depletion supernatant resulted from PI3K-p110 immunoprecipitation. These results indicated that arsenic increased PI3K-p85 which was free from PI3K-p110 binding. In addition, arsenic significantly increased interaction between IRS1 and PI3K-p85 but not PI3K-p110, suggesting that there may be a fraction of free PI3K-p85 interacting with IRS1. In vitro PI3K activity demonstrated that arsenic lowered PI3K activity in both basal and insulin-stimulated conditions. These results suggested that the increase in free PI3K-p85 by arsenic might compete with PI3K heterodimer for the same IRS1 binding site, in turn blocking the activation of its catalytic subunit, PI3K-p110. Taken together, our results provide additional insights into mechanisms underlying the impairment of neuronal insulin signaling by arsenic through the reduction of IR autophosphorylation, the increase in free PI3K-p85, and the impeding of PI3K activity.


Subject(s)
Arsenites/toxicity , Class Ia Phosphatidylinositol 3-Kinase/metabolism , Insulin/pharmacology , Neurons/drug effects , Sodium Compounds/toxicity , Antigens, CD/metabolism , Binding Sites , Cell Line, Tumor , Humans , Insulin Receptor Substrate Proteins/metabolism , Neurons/enzymology , Neurons/pathology , Phosphorylation , Protein Binding , Receptor, Insulin/agonists , Receptor, Insulin/metabolism , Signal Transduction
7.
Environ Toxicol Pharmacol ; 84: 103626, 2021 May.
Article in English | MEDLINE | ID: mdl-33621689

ABSTRACT

Arsenic is a metalloid that has been hypothesized to be an environmental risk factor for Alzheimer's disease (AD), a disease having hyperphosphorylated tau aggregate as a marker. The present study demonstrated that prolonged exposure to sodium arsenite at low micromolar range (1-10 µM) reduced Tau 1 (recognizing dephosphorylated tau at residues 189-207) and elevated pS202 tau in differentiated human neuroblastoma SH-SY5Y cells indicating that arsenic increases tau phosphorylation in neurons. Sodium arsenite elevated GSK3ß kinase activity, while GSK3 inhibitors, BIO, SB216763, and lithium, reversed the Tau 1 reduction by sodium arsenite. Additionally, sodium arsenite increased levels of active phosphorylation of ERK1/2, and inhibition of ERK1/2 by U0126 partially improved the Tau1 reduction. These results suggest that arsenic may cause tau hyperphosphorylation in neurons through the activation of GSK3 and ERK1/2. Furthermore, sodium arsenite augmented tau phosphorylation in the membrane and cytosolic fractions. Inductions of GSK3 activity by sodium arsenite treatment were observed in the membrane fraction, as evidenced by a reduction of ß-catenin, a protein signaled for degradation following phosphorylation by GSK3. An enhancement of ERK1/2 phosphorylation by sodium arsenite was also witnessed in the cytosol. Additionally, sodium arsenite increased insoluble tau aggregation. These results suggest that arsenic induces tau hyperphosphorylation in the membrane fraction which may lead to its redistribution from the membrane fraction to the cytosol, where it promotes neurofibrillary formation. Collectively, we demonstrate that prolonged arsenic exposure increases tau phosphorylation, partly through GSK3 and ERK1/2 activation, and insoluble tau aggregates, hence possibly contributing to the development of sporadic AD.


Subject(s)
Arsenic/toxicity , Environmental Pollutants/toxicity , Glycogen Synthase Kinase 3 beta/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , tau Proteins/metabolism , Cell Differentiation , Cell Line, Tumor , Cell Membrane/metabolism , Cytosol/metabolism , Humans , MAP Kinase Signaling System/drug effects , Phosphorylation/drug effects , Protein Aggregates/drug effects
8.
Toxicol Rep ; 6: 426-430, 2019.
Article in English | MEDLINE | ID: mdl-31193040

ABSTRACT

Andrographis paniculata is widely used in traditional herbal medicines for the treatment of common cold, fever and diarrhea, in many regions of Scandinavia and Asia, including Thailand. The pharmacological activities of A. paniculata are mainly attributed to active diterpenoids including 14-deoxyandrographolide, which is uniquely high in first true leaf ethanolic extract (FTLEE) of A. paniculata. In this study, the acute toxicity of the standardized FTLEE of A. paniculata was examined according to the OECD test guideline No. 420. Mice were divided into four groups of each sex and orally received the standardized FTLEE of A. paniculata (0, 300, 2000, or 5000 mg/kg BW). Post-treatment, body weight, signs of toxicity, and/or mortality were observed for 14 days. At Day 15, animals were euthanized, internal organs were observed grossly, and blood samples collected were subjected to hematology and clinical biochemistry analyses. The results showed that all treated animals survived and no apparent adverse effects were observed during the duration of the study. Gross necropsy observation revealed no lesion in any organ of all the standardized FTLEE-treated mice. Although significant alterations in BUN, lymphocytes, neutrophils, hematocrit and hemoglobin were observed, these alterations were not treatment-related toxic effects. Therefore, we concluded that a single oral administration of the standardized FTLEE of A. paniculata with an upper fixed dose of 5000 mg/kg BW has no significant acute toxicological effects.

9.
Environ Toxicol Pharmacol ; 66: 7-13, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30593951

ABSTRACT

Arsenic exposure has been linked to an impaired immune response and inflammation. Our study investigated the effects of sodium arsenite on host immune response and vascular inflammation during malarial infection. Mice were divided into three groups: control (C), Plasmodium berghei infection (I) and sodium arsenite exposure with Plasmodium berghei infection (As-I). The results showed that splenocyte proliferation stimulated by lipopolysaccharide (LPS) and pokeweed mitogen (PWM) was suppressed in the I group, and the suppression was more pronounced in the As-I group, suggesting that acquired immunity in infected mice was worsening following arsenic exposure. ICAM-1, an adhesion protein involved in parasite-infected red blood cell (iRBC) binding to endothelium, and HIF-1α, a hypoxia marker protein in the descending aorta, were increased in the As-I group compared to the I group. Collectively, our results suggest that arsenic may increase host susceptibility to malaria through suppression of B cell proliferation and enhancement of adhesion between iRBC and endothelium by increasing ICAM-1.


Subject(s)
Arsenites/toxicity , B-Lymphocytes/drug effects , Endothelium, Vascular/immunology , Malaria/immunology , Sodium Compounds/toxicity , Animals , Arsenites/blood , Arsenites/pharmacokinetics , B-Lymphocytes/immunology , Cell Proliferation/drug effects , Disease Models, Animal , Disease Susceptibility , Erythrocytes/immunology , Inflammation/immunology , Intercellular Adhesion Molecule-1/immunology , Male , Mice , Plasmodium berghei , Sodium Compounds/blood , Sodium Compounds/pharmacokinetics , Tissue Distribution
10.
Int J Oncol ; 53(1): 177-188, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29693152

ABSTRACT

Cholangiocarcinoma (CCA) is a malignant tumor of the biliary epithelium associated with Opisthorchis viverrini, primary sclerosing cholangitis and hepatitis viral infection. In the global population, men have higher incidence rates for CCA than women; thus, a gender disparity in the progression of chronic inflammation of the biliary duct leading to malignancy may involve the effects of estrogen (E2). Genistein (GE), a prominent phytoestrogen found in soy products, is an estrogen receptor ß (ERß) agonist and a tyrosine kinase inhibitor. The present study investigated the effects of GE on the growth of CCA cells by cell viability assay. The effects on signaling proteins were detected by western blot analysis and ELISA. Gene expression was examined by RT-qPCR. Two human intrahepatic CCA cell lines, HuCCA­1 and RMCCA­1, were utilized. GE (50­200 µM) reduced the viability of the two cell lines, and also inhibited the activation of epidermal growth factor receptor (EGFR) and AKT, as evidenced by decreasing protein levels of phosphorylated (p)-EGFR (Tyr1173) and p­AKT (Ser473), respectively. GE altered the mitogen­activated protein kinase signaling cascade by mediating decreased protein levels of p­p38 and increased protein levels of p­ERK1/2. GE significantly decreased the levels of interleukin 6 (IL6) and induced the expression of inducible nitric oxide synthase (iNOS). GE also downregulated the expression of p­ERα (Ser118) protein and ERα mRNA levels. Finally, GE induced the downregulation of the protein levels of ERß. Of note, E2 deprivation potentiated the GE-induced reduction of p­EGFR (Tyr1173) and total AKT proteins and production of IL6, and mediated the downregulation of GE-induced iNOS protein. In conclusion, GE inhibited the growth of human CCA cell lines by reducing the activation of EGFR and AKT, and by attenuating the production of IL6. E2 and ER were also involved in the growth-inhibitory effect of GE in CCA cells.


Subject(s)
Cholangiocarcinoma/drug therapy , ErbB Receptors/genetics , Genistein/administration & dosage , Interleukin-6/genetics , Oncogene Protein v-akt/genetics , Cell Line, Tumor , Cell Survival/drug effects , Cholangiocarcinoma/genetics , Cholangiocarcinoma/pathology , ErbB Receptors/antagonists & inhibitors , Estrogen Receptor beta/agonists , Estrogen Receptor beta/genetics , Estrogens/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Oncogene Protein v-akt/antagonists & inhibitors , Phosphorylation , Signal Transduction/drug effects
11.
Neurotoxicology ; 66: 22-31, 2018 05.
Article in English | MEDLINE | ID: mdl-29526746

ABSTRACT

A strong correlation between chronic arsenic exposure and neuropsychological disorders leads to a growing concern about a potential risk of arsenic related neurodegeneration. Evidently, brain insulin signaling contributes to physiological effects, including energy homeostasis, and learning and memory. Arsenic has been shown to impair insulin signaling in adipocytes and myocytes, however, this impairment has not yet been explored in neurons. Here we showed that NaAsO2 caused significant reduction in basal levels of glucose, plasma membrane glucose transporter, GLUT 3 and Akt phosphorylation in differentiated human neuroblastoma SH-SY5Y cells. NaAsO2 significantly decreased insulin-mediated glucose uptake, as well as GLUT1 and 3 membrane translocation. Furthermore, the ability of insulin to increase Akt phosphorylation, a well-recognized insulin signaling response, was significantly lessened by NaAsO2 treatment. In addition, the classical tyrosine phosphorylation response of insulin was reduced by NaAsO2, as evidenced by reduction of insulin-induced tyrosine phosphorylation of insulin receptor (IR) and insulin receptor substrate-1(IRS-1). Moreover, NaAsO2 lowered the ratio of p110, a catalytic subunit to p85, a regulatory subunit of PI3K causing an imbalance between p110 and p85, the conditions reported to contribute to insulin sensitivity. Additionally, increment of IRS-1 interaction with GSK3ß, and p85-PI3K were observed in NaAsO2 treated cells. These molecular modulations may be mechanistically attributed to neuronal insulin signaling impairment by arsenic.


Subject(s)
Arsenic/toxicity , Insulin/metabolism , Cell Line, Tumor , Glucose/metabolism , Glucose Transporter Type 3/metabolism , Humans , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Insulin/metabolism , Signal Transduction
12.
Toxicol In Vitro ; 42: 123-129, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28414160

ABSTRACT

Paraquat (PQ) is a bipyridyl derivative herbicide known to cause lung toxicity partly through induction of apoptosis. Here we demonstrated that PQ caused apoptosis in A549 cells. PQ increased cleavage of caspase-8 and Bid, indicating caspase-8 activation and truncated Bid, the two key mediators of extrinsic apoptosis. Additionally, PQ treatment caused an increase in DR5 (death receptor-5) and caspase-8 interaction, indicating formation of DISC (death-inducing signaling complex). These results indicate that PQ induces apoptosis through extrinsic pathway in A549 cells. Moreover, PQ drastically increased DR5 expression and membrane localization. Furthermore, PQ caused prominent concentration dependent reductions of DDX3 (the DEAD box protein-3) and GSK3 (glycogen synthase kinase-3) which can associate with DR5 and prevent DISC formation. Additionally, PQ decreased DR5-DDX3 interaction, suggesting a reduction of DDX3/GSK3 anti-apoptotic complex. Inhibition of GSK3, which is known to promote extrinsic apoptosis by its pharmacological inhibitor, BIO accentuated PQ-induced apoptosis. Moreover, GSK3 inhibition caused a further decrease in PQ-reduced DR5-DDX3 interaction. Taken together, these results suggest that PQ may induce extrinsic pathway of apoptosis in A549 cells through upregulation of DR5 and repression of anti-apoptotic proteins, DDX3/GSK3 leading to reduction of anti-apoptotic complex.


Subject(s)
Herbicides/toxicity , Paraquat/toxicity , A549 Cells , Apoptosis/drug effects , DEAD-box RNA Helicases/metabolism , Glycogen Synthase Kinase 3/metabolism , Humans , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism
13.
J Appl Toxicol ; 36(6): 790-801, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26234195

ABSTRACT

Perfluorooctane sulfonate (PFOS) and perfluorooctanoic acid (PFOA) are the two most popular surfactants among perfluorinated compounds (PFCs), with a wide range of uses. Growing evidence suggests that PFCs have the potential to interfere with estrogen homeostasis, posing a risk of endocrine-disrupting effects. This in vitro study aimed to investigate the estrogenic effect of these compounds on T47D hormone-dependent breast cancer cells. PFOS and PFOA (10(-12) to 10(-4) M) were not able to induce estrogen response element (ERE) activation in the ERE luciferase reporter assay. The ERE activation was induced when the cells were co-incubated with PFOS (10(-10) to 10(-7) M) or PFOA (10(-9) to 10(-7) M) and 1 nM of 17ß-estradiol (E2). PFOS and PFOA did not modulate the expression of estrogen-responsive genes, including progesterone (PR) and trefoil factor (pS2), but these compounds enhanced the effect of E2-induced pS2 gene expression. Neither PFOS nor PFOA affected T47D cell viability at any of the tested concentrations. In contrast, co-exposure with PFOS or PFOA and E2 resulted in an increase of E2-induced cell viability, but no effect was found with 10 ng ml(-1) EGF co-exposure. Both compounds also intensified E2-dependent growth in the proliferation assay. ERK1/2 phosphorylation was increased by co-exposure with PFOS or PFOA and E2, but not with EGF. Collectively, this study shows that PFOS and PFOA did not possess estrogenic activity, but they enhanced the effects of E2 on estrogen-responsive gene expression, ERK1/2 activation and the growth of the hormone-deprived T47D cells. Copyright © 2015 John Wiley & Sons, Ltd.


Subject(s)
Alkanesulfonic Acids/toxicity , Breast Neoplasms/chemically induced , Caprylates/toxicity , Endocrine Disruptors/toxicity , Estradiol/agonists , Estrogens/agonists , Fluorocarbons/toxicity , Surface-Active Agents/toxicity , Alkanesulfonic Acids/antagonists & inhibitors , Butadienes/pharmacology , Caprylates/antagonists & inhibitors , Carcinogens, Environmental/chemistry , Carcinogens, Environmental/toxicity , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Endocrine Disruptors/chemistry , Estradiol/pharmacology , Estrogens/pharmacology , Female , Fluorocarbons/antagonists & inhibitors , Gene Expression Regulation, Neoplastic/drug effects , Genes, Reporter/drug effects , Humans , MAP Kinase Signaling System/drug effects , Neoplasm Proteins/agonists , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Nitriles/pharmacology , Osmolar Concentration , Protein Kinase Inhibitors/pharmacology , Response Elements/drug effects , Surface-Active Agents/chemistry , Trefoil Factor-1/agonists , Trefoil Factor-1/genetics , Trefoil Factor-1/metabolism
14.
Environ Toxicol ; 31(9): 1133-46, 2016 Sep.
Article in English | MEDLINE | ID: mdl-25728338

ABSTRACT

Arsenic (As) is considered a major environmental health threat worldwide due to its widespread contamination in drinking water. Recent studies reported that arsenic is a potential xenoestrogen as it interfered with the action of estrogen (E2) and estrogen receptor (ER) signaling. The present study investigated the effects of sodium arsenite (NaAsO2 ) on estrogen signaling in human breast cancer cells. The results demonstrated that NaAsO2 dose-dependently increased viability of hormone-dependent breast cancer MCF-7 and T47D cells expressing both ERα and ERß but not hormone-independent MDA-MB-231 cells expressing ERß. These suggested ERα contribution to NaAsO2 -stimulated breast cancer cells growth. NaAsO2 induced down-regulation of ERα but up-regulation of ERß protein expressions in T47D cells. Moreover, NaAsO2 dose-dependently inhibited E2-induced ER transcriptional activity as it decreased E2-mediated ERE-luciferase transcription activation and PgR mRNA transcription but increased pS2 mRNA transcription. However, NaAsO2 induced both rapid and sustained activation of ERK1/2 and increased in phosphorylation of ERα at serine 118 residue, c-fos and c-myc protein expressions. These results indicated that NaAsO2 interferes the genomic estrogen-signaling pathway but induces activation of a rapid nongenomic signal transduction through ERK1/2 pathway which may contribute to its proliferative effect on hormone-dependent breast cancer cells. © 2015 Wiley Periodicals, Inc. Environ Toxicol 31: 1133-1146, 2016.


Subject(s)
Arsenites/toxicity , Estrogen Receptor alpha/metabolism , Signal Transduction/drug effects , Sodium Compounds/toxicity , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Down-Regulation/drug effects , Estradiol/pharmacology , Estrogen Receptor beta/metabolism , Female , Humans , MCF-7 Cells , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation/drug effects , Presenilin-2/genetics , Presenilin-2/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-myc/metabolism , RNA, Messenger/metabolism , Transcriptional Activation/drug effects , Up-Regulation/drug effects
15.
Toxicology ; 338: 117-29, 2015 Dec 02.
Article in English | MEDLINE | ID: mdl-26514924

ABSTRACT

Aside from the effects on neuronal cholinergic system, epidemiological studies suggest an association between chlorpyrifos (CPF) exposure and cancer risk. This in vitro study examined the effects of CPF and its toxic metabolite, chlorpyrifos oxon (CPF-O), on the growth of human colorectal adenocarcinoma H508, colorectal adenocarcinoma HT-29, normal colon epithelial CCD841, liver hepatocellular carcinoma HepG2, and normal liver hepatocyte THLE-3 cells. The results showed that CPF (5-100 µM) concentration-dependently increased viability of H508 and CCD841 cells in serum-free conditions. This increasing trend was not found in HT-29, HepG2 and THLE-3 cells. In contrast, CPF-O (50-100 µM) reduced the viability of all cell lines. Cell cycle analysis showed the induction of cells in the S phase, and EdU incorporation assay revealed the induction of DNA synthesis in CPF-treated H508 cells indicating that CPF promotes cell cycle progression. Despite the observation of acetylcholinesterase activity inhibition and reactive oxygen species (ROS) generation, atropine (a non-selective muscarinic acetylcholine receptor antagonist) and N-acetylcysteine (a potent antioxidant) failed to inhibit the growth-promoting effect of CPF. CPF increased the phosphorylation of epidermal growth factor receptor (EGFR) and its downstream effector, extracellular signal regulated kinase (ERK1/2), in H508 cells. AG-1478 (a specific EGFR tyrosine kinase inhibitor) and U0126 (a specific MEK inhibitor) completely mitigated the growth promoting effect of CPF. Altogether, these results suggest that EGFR/ERK1/2 signaling pathway but not cholinergic pathway involves in CPF-induced colorectal adenocarcinoma H508 cell growth.


Subject(s)
Adenocarcinoma/enzymology , Cell Proliferation/drug effects , Chlorpyrifos/analogs & derivatives , Colorectal Neoplasms/enzymology , ErbB Receptors/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Pesticides/toxicity , Signal Transduction/drug effects , Acetylcholinesterase/metabolism , Adenocarcinoma/pathology , Antioxidants/pharmacology , Cell Cycle/drug effects , Cell Survival/drug effects , Chlorpyrifos/toxicity , Colorectal Neoplasms/pathology , Dose-Response Relationship, Drug , ErbB Receptors/antagonists & inhibitors , GPI-Linked Proteins/antagonists & inhibitors , GPI-Linked Proteins/metabolism , HT29 Cells , Hep G2 Cells , Humans , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Muscarinic Antagonists/pharmacology , Oxidative Stress/drug effects , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Reactive Oxygen Species/metabolism , Time Factors
16.
Toxicol Lett ; 236(3): 138-44, 2015 Aug 05.
Article in English | MEDLINE | ID: mdl-25982963

ABSTRACT

Arsenic neurotoxicity has a broad range of adverse effects on human health, which are induced in part by inhibition of neurite outgrowth. Insulin has been reported to promote neurite extension. The present study investigated whether insulin can protect neurons from impaired neurite outgrowth induced by arsenic, and examined the signaling pathway involved in this action. The study demonstrated that NaAsO2 caused inhibition of neurite outgrowth in differentiated SH-SY5Y cells indicating its neurotoxicity. This inhibitory effect of NaAsO2 was attenuated by insulin. It was found that blocking PI3K or Akt by selective inhibitors canceled the protective effect of insulin against NaAsO2-induced neurite outgrowth impairment suggesting the essential role of active PI3K and Akt in insulin's protective action. Inhibition of GSK3, which mimics an effect of insulin stimulation, had no effect on the impairment of neurite outgrowth by NaAsO2 implying that the insulin protective action is probably not due to its mediation of GSK3 inhibition ability. Moreover, NaAsO2 decreased the Akt activity, as it caused reduction in Akt phosphorylation, and downregulated expression of SIRT1. Additionally, the reduction of these signals by NaAsO2 was attenuated by insulin. Taken together, these results show that insulin attenuates arsenic-induced neurite outgrowth impairment possibly via activation of PI3K/Akt/SIRT1 signaling, and arsenic may exert neurite outgrowth inhibition through a mechanism involving reduction of signaling molecules downstream from insulin, PI3K/Akt/SIRT1. Our findings raise the possibility of using insulin to combat arsenic neurotoxicity.


Subject(s)
Arsenites/toxicity , Insulin/pharmacology , Neurites/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Sirtuin 1/metabolism , Sodium Compounds/toxicity , Cell Line, Tumor , Gene Expression Regulation/drug effects , Humans , Neurites/physiology , Neuroblastoma , Phosphatidylinositol 3-Kinases/genetics , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/genetics , Sirtuin 1/genetics
17.
Int J Oncol ; 46(6): 2317-26, 2015.
Article in English | MEDLINE | ID: mdl-25815516

ABSTRACT

Cholangiocarcinoma (CCA) is a malignant cancer of the biliary tract and its occurrence is associated with chronic cholestasis which causes an elevation of bile acids in the liver and bile duct. The present study aimed to investigate the role and mechanistic effect of bile acids on the CCA cell growth. Intrahepatic CCA cell lines, RMCCA-1 and HuCCA-1, were treated with bile acids and their metabolites to determine the growth promoting effect. Cell viability, cell cycle analysis, EdU incorporation assays were conducted. Intracellular signaling proteins were detected by western immunoblotting. Among eleven forms of bile acids and their metabolites, only taurolithocholic acid (TLCA) concentration dependently (1-40 µM) increased the cell viability of RMCCA-1, but not HuCCA-1 cells. The cell cycle analysis showed induction of cells in the S phase and the EdU incorporation assay revealed induction of DNA synthesis in the TLCA-treated RMCCA-1 cells. Moreover, TLCA increased the phosphorylation of EGFR, ERK 1/2 and also increased the expression of cyclin D1 in RMCCA-1 cells. Furthermore, TLCA-induced RMCCA-1 cell growth could be inhibited by atropine, a non-selective muscarinic acetylcholine receptor (mAChR) antagonist, AG 1478, a specific EGFR inhibitor, or U 0126, a specific MEK 1/2 inhibitor. These results suggest that TLCA induces CCA cell growth via mAChR and EGFR/EKR1/2 signaling pathway. Moreover, the functional presence of cholinergic system plays a certain role in TLCA-induced CCA cell growth.


Subject(s)
Bile Acids and Salts/adverse effects , Bile Duct Neoplasms/pathology , Cholangiocarcinoma/pathology , MAP Kinase Signaling System/drug effects , Taurolithocholic Acid/adverse effects , Bile Acids and Salts/pharmacology , Bile Duct Neoplasms/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cholangiocarcinoma/metabolism , Cyclin D1/metabolism , ErbB Receptors/metabolism , Humans , Phosphorylation , Receptors, Muscarinic , Taurolithocholic Acid/pharmacology
18.
Planta Med ; 80(7): 533-43, 2014 May.
Article in English | MEDLINE | ID: mdl-24782229

ABSTRACT

Andrographis paniculata is an important herbal medicine widely used in several Asian countries for the treatment of various diseases due to its broad range of pharmacological activities. The present study reports that A. paniculata extracts potently inhibit the growth of liver (HepG2 and SK-Hep1) and bile duct (HuCCA-1 and RMCCA-1) cancer cells. A. paniculata extracts with different contents of major diterpenoids, including andrographolide, 14-deoxy-11,12-didehydroandrographolide, neoandrographolide, and 14-deoxyandrographolide, exhibited a different potency of growth inhibition. The ethanolic extract of A. paniculata at the first true leaf stage, which contained a high amount of 14-deoxyandrographolide but a low amount of andrographolide, showed a cytotoxic effect to cancer cells about 4 times higher than the water extract of A. paniculata at the mature leaf stage, which contained a high amount of andrographolide but a low amount of 14-deoxyandrographolide. Andrographolide, not 14-deoxy-11,12-didehydroandrographolide, neoandrographolide, or 14-deoxyandrographolide, possessed potent cytotoxic activity against the growth of liver and bile duct cancer cells. The cytotoxic effect of the water extract of A. paniculata at the mature leaf stage could be explained by the present amount of andrographolide, while the cytotoxic effect of the ethanolic extract of A. paniculata at the first true leaf stage could not. HuCCA-1 cells showed more sensitivity to A. paniculata extracts and andrographolide than RMCCA-1 cells. Furthermore, the ethanolic extract of A. paniculata at the first true leaf stage increased cell cycle arrest at the G0/G1 and G2/M phases, and induced apoptosis in both HuCCA-1 and RMCCA-1 cells. The expressions of cyclin-D1, Bcl-2, and the inactive proenzyme form of caspase-3 were reduced by the ethanolic extract of A. paniculata in the first true leaf stage treatment, while a proapoptotic protein Bax was increased. The cleavage of poly (ADP-ribose) polymerase was also found in the ethanolic extract of A. paniculata in the first true leaf stage treatment. This study suggests that A. paniculata could be a promising herbal plant for the alternative treatment of intrahepatic cholangiocarcinoma.


Subject(s)
Andrographis/chemistry , Bile Duct Neoplasms/drug therapy , Cholangiocarcinoma/drug therapy , Diterpenes/therapeutic use , Phytotherapy , Plant Extracts/therapeutic use , Apoptosis/drug effects , Bile Ducts, Intrahepatic , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Diterpenes/chemistry , Diterpenes/isolation & purification , Humans , Inhibitory Concentration 50 , Plant Extracts/chemistry , Plant Extracts/isolation & purification , Plants, Medicinal
19.
Mitochondrion ; 14(1): 54-63, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24316184

ABSTRACT

Mitochondrial biogenesis, a mitochondrial growth and division process, is crucial for adaptation to metabolic stress. The present study demonstrated that treatment with a specific inhibitor of GSK3, SB216763, attenuated induction of mitochondrial biogenesis by a glycolysis inhibitor, 2-deoxyglucose (2-DG), without affecting this biogenesis at basal condition. Additionally, overexpression of WT-GSK3ß promoted whereas GSK3ß-KD attenuated 2-DG-induced mitochondrial protein expression. The mitochondrial biogenesis attenuation by GSK3 inhibitor was not due to inhibition of protein degradation. Furthermore, GSK3 inhibition further reduced transcription of mitochondrial (COXII), but not nuclear (VDAC) gene by 2-DG suggesting its participation in 2-DG-induced mitochondrial transcription. Together, our results show that GSK3 regulates mitochondrial biogenesis induced by glycolysis inhibition.


Subject(s)
Deoxyglucose/metabolism , Glycogen Synthase Kinase 3/metabolism , Mitochondrial Turnover , Neurons/enzymology , Neurons/physiology , Cell Line, Tumor , Enzyme Inhibitors/metabolism , Gene Expression , Humans , Indoles/metabolism , Maleimides/metabolism , Neurons/metabolism
20.
Environ Toxicol ; 29(6): 672-8, 2014 Jun.
Article in English | MEDLINE | ID: mdl-22859221

ABSTRACT

Arsenic is a widespread contaminant in the environment especially in drinking water. Although it is a known carcinogen in human, the mechanism by which arsenic induces carcinogenesis is not well understood. Among several effects of arsenic, it has been suggested that arsenic-induced vascular endothelial growth factor (VEGF) expression plays a critical role in arsenic carcinogenesis. In the present study, we demonstrated that arsenite induced VEGF expression in neuroblastoma SH-SY5Y cells without induction of HIF-1α, a well-known transcriptional activator for VEGF suggesting that arsenite-induced VEGF expression in SH-SY5Y cells may not require HIF-1α activation. It has been reported that VEGF expression is regulated by multiple transcription factors including ß-catenin. We therefore investigated whether ß-catenin was involved in arsenite-induced VEGF expression in SH-SY5Y cells. Treatment of arsenite caused ß-catenin accumulation in the nucleus. Additionally, arsenite treatment decreased the activity of GSK3, an enzyme that phosphorylates and targets ß-catenin for degradation by proteasome, without activation of its upstream kinase, Akt. Inhibition of PI3K/Akt which negatively regulates GSK3 activity by LY294002 resulted in a decrease in arsenite-mediated ß-catenin nuclear accumulation, and VEGF expression. These results suggested that ß-catenin plays a role in arsenite-induced VEGF in SH-SY5Y cells, and the induction of ß-catenin by arsenite is mediated by inhibition of GSK3 without activating its upstream kinase Akt.


Subject(s)
Arsenites/toxicity , Neuroblastoma/metabolism , Vascular Endothelial Growth Factor A/metabolism , beta Catenin/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Chromones/pharmacology , Glycogen Synthase Kinase 3/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Morpholines/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...